Skip to main content

Full text of "USPTO Patents Application 09993647"

See other formats


(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 



(19) World Intellectual Property Organization 

International Bureau 

(43) International Publication Date 
2 August 2001 (02.08.2001) 




PCT 



lllllllllilllllllllllllllllli 

(10) International Publication Number 

WO 01/54723 Al 



(51) International Patent Ciassification^: A61K 39/395, (74) Agent: SOMERVILLE, Deborah, A.; Kenyon & 
39/00, 39/38, GOIN 33/53 Kenyon, One Broadway, New York, NY 10004 (US). 



(21) International Application Number: PCT/USO 1/02839 

(22) International Filing Date: 29 January 2001 (29.0L2001) 



(25) Filing Language: 

(26) Publication Language: 



English 



English 



(30) Priority Data: 

60/178,791 
09/539,692 



28 January 2000 (28.01 .2000) US 
31 March 2000 (31.03.2000) US 



(71) Applicant (for all designated States except BB, US): SUN- 
NYBROOK HEALTH SCIENCE CENTER [CA/CA]; 
2075 Bayview Avenue, North York, Ontario M4N 3M5 
(CA). 

(71) Applicant (for BB only): IMCLONE SYSTEMS IN- 
CORPORATED [US/US]; 180 Varick Street, New York, 
NY 10014 (US). 

(72) Inventor; and 

(75) Inventor/Applicant (for US only): KERBEL, Robert 

[CA/CA]; 48 Bennington Heights Drive, Toronto, Ontario 
M4G 1A9 (CA). 



(81) Designated States (national): AE, AG, AL, AM, AT, AU, 
AZ, BA, BB, BG, BR, BY, BZ, CA, CH, CN, CR, CU, CZ, 
DE, DK, DM, DZ, EE, ES, FI, GB, GD, GE, GH, GM, HR, 
HU, ID, XL, IN, IS, JP, KE, KG, KP, KR, KZ, LC, LK, LR, 
LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX, MZ, 
NO, NZ, PL, PT, RO, RU, SD, SE, SG, SI, SK, SL, TJ, TM, 
TR, TT, TZ, UA, UG, US, UZ, VN, YU, ZA, ZW. 

(84) Designated States (regional): ARIPO patent (GH, GM, 
KE, LS, MW, MZ, SD, SL, SZ, TZ, UG, ZW), Eurasian 
patent (AM, AZ, BY, KG, KZ, MD, RU, TJ, TM), European 
patent (AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, 
IT, LU, MC, NL, PT, SE, TR), OAPI patent (BF, BJ, CF, 
CG, CI, CM, GA, GN, GW, ML, MR, NE, SN, TD, TG). 

Published: 

— with international search report 

— before the expiration of the time limit for amending the 
claims and to be republished in the event of receipt of 
amendments 

For two-letter codes and other abbreviations, refer to the "Guid- 
ance Notes on Codes and Abbreviations" appearing at the begin- 
ning of each regular issue of the PCT Gazette. 



f<| (54) Title: THERAPEUTIC METHOD FOR REDUCING ANGIOGENESIS 



IT) 



o 



(57) Abstract: A method of controlling or treating an angiogenic dependent condition in a mammal, preferably in a human by ad- 
ministering an anti-angiogenic molecule such as an angiogenesis growth factor antagonist, and a chemotherapeutic agent in amounts 
and frequencies effective, in combination, to produce a regression or arrest of said condition while minimizing or preventing signif- 
icant toxicity of the chemotherapeutic agent. Also a kit for controlling or treating an angiogenic dependent condition in a mammal, 
preferably in a human, comprising an anti-angiogenic molecule, such as an angiogenesis growth factor antagonist, and a chemothera- 
peutic agent in amounts effective, in combination, to produce a regression or arrest of said condition while minimizing or preventing 
significant toxicity of the chemotherapeutic agent. 



wo 01/54723 



PCT/USOl/02839 



THERAPEUTIC METHOD FOR REDUCING ANGIOGENESIS 

The present application claims the benefit of priority from U.S. Provisional 
Application No. 60/178791, filed on January 28, 2000, which is hereby incorporated in its 
entirety by reference. 

Field of the Invention 

The present invention relates to the inhibition or prevention of angiogenesisas a means 
to control or treat an angiogenic dependent condition, a condition characterized by, or 
dependent upon, blood vessel proliferation. The invention further relates to the use of an anti- 
angiogenic molecule in combination with a chemotherapeutic agent. 

Background of the Invention 

Angiogenesis is a highly complex process of developing new blood vessels that involves 
the proliferation and migration of, and tissue infiltration by capillar)^ endothelial cells fi-om pre- 
existing blood vessels, cell assembly into tubular stRic^iire?. ' oining of newly forming tubular 
assemblies to closed-circuit vascular systems, and maturation of newly formed capillary vessels. 
The molecular bases of many of these aspects are still not understood. 

Angiogenesis is important in normal physiological processes including embryonic 
development, follicular growth, and wound healing, as well as in pathological conditions such as 
tumor growth and in non-neoplastic diseases involving abnonnal neovascularization, including 
neovascular glaucoma (Folkman, J. and Klagsbrun, M. Science 235:442-447 (1987). Other 
disease states include but are not limited to, neoplastic diseases, mcluding but not limited to solid 
tumors, autoimmune diseases and collagen vascular diseases such as, for example, rheumatoid 
arthritis, and ophthalmalogical conditions such as diabetic retinopathy, retrolental fibroplasia and 
neovascular glaucoma. Conditions or diseases to which persistent or uncontrolled angiogenesis 
contribute have been termed angiogenic dependent or angiogenic associated diseases. 

One means of controlling such diseases and pathological conditions comprises restricting 
the blood supply to those cells involved in mediating or causing the disease or condition. For 
example, in the case of neoplastic disease, solid tumors develop to a size of about a few 
millimeters, and further growth is not possible, absent angiogenesis within the tumor. In the past. 



wo 01/54723 



PCT/USOl/02839 



strategies to limit the blood supply to tumors have included occluding blood vessels supplying 
portions of organs in which tumors are present. Such approaches require the site of the tumor to 
be identified and are generally limited to treatment to a single site, or small number of sites. An 
additional disadvantage of direct mechanical restriction of a blood supply is that collateral blood 
vessels develop, often quite rapidly, restoring the blood supply to the tumor. 

Other approaches have focused on the modulation of factors that are involved in the 
regulation of angiogenesis. While usually quiescent, vascular endothelial proliferation is highly 
regulated, even during angiogenesis. Examples of factors that have been implicated as possible 
regulators of angiogenesis in vivo include, but are not limited to, transforming growth factor beta 
(TGFp), acidic and basic fibroblast growth factor (aFGF and bFGF), platelet derived growth 
factor (PDGF), and vascular endothelial growth factor (VEGF) (Klagsbrun, M. and D'Amore, P. 

(1991) Annual Rev. Physiol. 53: 217-239). 

One growth factor of particular interest is VEGF. An endothelial-cell specific mitogen, 
VEGF acts as an angiogenesis inducer by specifically promoting the proliferation of endothelial 
ceils. It is a homodimeric glycoprotein consisting of two 23 kD subunits with structural similarity 
to PDGF, Four different monomeric isoforms of VEGF resulting fi-om altemative splicing of 
n-iRN.A ; J been identi. ;ed ^hese include two membrane bound forms (VEGF206 and VEGFjgg) 
^. : ;v e toims • ' ' i and VEGF, 3,). VEGF,^,s is the most abundant isoform in all 

huniun -Mies except . - . 

\ LGF is expres^-j.. - embryonic tissues (Breier et al.. Development (Camb.) 1 14:521 

(1992) ), macrophages, and proliferating epidermal keratinocytes during wound healing (Brown et 
al., J, Exp. Med., 176:1375 (1992)), and may be responsible for tissue edema associated with 
inflammation (Fenrara et al., Endocr. Rev. 13:18 (1992)). In situ hybridization studies have 
demonstrated high levels of VEGF expression in a number of human tumor lines including 
glioblastoma multiforme, hemangioblastoma, other central nervous system neoplasms and AIDS- 
associated Kaposi^s sarcoma (Plate, K. et al. (1992) Nature 359: 845-848; Plate, K. et al. (1993) 
Cancer Res. 53: 5822-5827; Berkman, R. et al. (1993) J. Clin. Invest. 91: 153-159; Nakamura, S. 
et al. (1992) AIDS Weekly, 13(1)). High levels of VEGF also have been reported in hypoxia 
induced angiogenesis (Shweiki, D. et al. (1992) Nature 359: 843-845). 

VEGF mediates its biological effect through high affinity VEGF receptors which are 
selectively expressed on endothelial cells during, for example, embryogenesis (Millauer, B., et al. 

(1993) Cell 72: 835-846) and tumor formation. VEGF receptors typically are class III receptor- 
type tyrosine kinases characterized by having several, typically 5 or 7, immunoglobulin-like loops 



wo 01/54723 



PCT/USOl/02839 



in their amino-terminal extracellialar receptor ligand-binding domains (Kaipainen et al., J. Exp. 
Med. 178:2077-2088 (1993)). The other two regions include a transmembrane region and a 
carboxy-terminal intracellular catalytic domain interrupted by an insertion of hydrophilic 
interkinase sequences of variable lengths, called the kinase insert domain (Terman et al,. 
Oncogene 6:1677-1683 (1991)). VEGF receptors include flt-1, sequenced by Shibuya M. et al.. 
Oncogene 5, 519-524 (1990); flk-1, sequenced by Matdiews W. et al. Proc. Natl. Acad. Sci. USA, 
88:9026-9030 (1991) and KDR the human homologue of flk-1, described in PCT/US92/01300, 
filed Februaiy 20, 1992, and in Temian et al.. Oncogene 6:1677-1683 (1991). 

High levels of flk-1 are expressed by endothelial cells that infiltrate gliomas (Plate, K. et 
al., (1992) Nature 359: 845-848), and are specifically upregulated by VEGF produced by human 
glioblastomas (Plate, K. et al. (1993) Cancer Res. 53: 5822-5827). The finding of high levels of 
flk-1 expression in glioblastoma associated endothelial cells (GAEC) suggests that receptor 
activity is induced during tumor fomiation, since flk- 1 transcripts are barely detectable in normal 
brain endothelial cells. This upregulation is confined to the vascular endothelial cells in close 
proximity to the tumor. Blocking VEGF activity with neutralizing anti-VEGF monoclonal 
antibodies (mAbs) results in inhibition of the growth of human tumor xenografts in nude mice 
(Kim. K. et al. (1993) Nature 362: 841-844), suggesting a direct role for VEGF in tumor-related 
angiogener:". 

Various chemotherapeutic drugs also have been showii to block fiinctions of activated, 
dividing endothelial cells critical to angiogenesis, or to kill such cells. Such collateral damaging 
effects on a genetically stable normal host cell, in addition to the chemotherapeutic agent's effect 
upon the tumor cells, contribute significantly to the in vivo anti-tumor efficacy of chemotherapy . 
However, the standard use of chemotherapeutic agents has obvious undesirable side-effects upon 
the normal cells of patients, limiting its use. Administration of chemotherapeutic agents in their 
usual doses and at usual dosage fi-equencies are commonly associated with side-effects, including, 
but not limited to, myelosuppression, neurotoxicity, cardiotoxicity, alopecia, nausea and vomiting, 
nephrotoxicity, and gastrointestinal toxicity. Further, patients' tumors often also develop 
resistance to the chemotherapeutic agents after initial exposure to the drugs. 

A desirable method and composition for controlling angiogenesis should be well 
tolerated, have few or no side-effects, and prevent new vessel formation at sites of disease 
without interfering with required physiologic angiogenesis in normal sites. It should be 
effective and, in the case of neoplastic disease, overcome the problem of the development of 
drug resistance by tumor cells. In so doing, it should permit targeted therapy without the 



wo 01/54723 



PCT/USOl/02839 



accurate identification of all disease sites. The present invention addresses many of the 
problems with existing materials and methods. 

SUMMARY OF THE INVENTION 

5 

The present invention provides a method of treating an angiogenic dependent 
condition in a mammal comprising administering an anti-angiogenic molecule and a 
chemotherapeutic agent to the mammal, in an amount and frequency effective, in 
combination, to produce a regression or arrest of the condition without significant toxicity 

10 from the chemotherapeutic agent. The angiogenic dependent condition may be selected from 
the group consisting of neoplasm, collagen-vascular disease or auto-immune disease, 
including a solid tumor neoplasm, including breast carcinoma, lung carcinoma, prostate 
carcinoma, colon carcinoma, prostate carcinoma, ovarian carcinoma, neuroblastoma, central 
nervous system tumor, neuroblastoma, glioblastoma multiforme or melanoma. The mammal 

15 receiving the treatment is preferably a human. 

The anti-angiogenic molecules inhibit the action of a vascular endothelium survival 
factor, which include receptors and their ligands. Vascular endothelium survival factors 
include /.c. * ^.eluding angiogenic growth factors such as VFC receptor, including flk- 
1/KDR receptor, or flt-4 receptor and VEGF. Examples of other vascular endothelial survival 

20 factors are integnn ayp., ttvP:. Hgand, Tie2/tek ligand. Tie2/tek. endoglin ligand, endoglin, 
neuropilin ligand, neuropilin, thrombospondin ligand, thrombospondin, PDGFa, PDGFa 
receptor, PDGFp, PDGFp receptor, aFGF, aFGF receptor, bFGF, bFGF receptor, TGFp, 
TGFp receptor, EGF, EGF receptor, angiostatin, angiostatin receptor, angiopoetin, 
angiopoeitin receptor, PLGF, PLGF receptor, VPF, or VPF receptor. Optionally, the ligand is 

25 selected from the group consisting of VEGF (VEGF-A), VEGF-B, VEGF-C, or VEGF-D. 
The anti-angiogenic molecule may be selected from the group consisting of antibody, 
antibody fragment, small molecule or peptide. 

Preferred embodiments of the present invention include antibodies selected from the 
group consisting of mouse antibody, rat antibody, chimeric antibody, humanized antibody or 

30 human antibody. A preferred antibody is IMC-lCl 1 . 

Preferably, IMC-lCll is administered in a dose of from about 5 mg/m- to about 700 
mg/nr about daily to about every 7 days, more preferably a dose of from about 7.5 mg/m- to 
about 225 mg/m-, about twice per week. Optionally, the IMC- 1 CI 1 is administered at a dose 

4 



wo 01/54723 



PCT/USOl/02839 



and frequency sufficient to substantially saturate the VEGF receptor. Optionally, the anti- 
angiogenic molecule is administered in a dose and frequency sufficient to substantially 
saturate the target of the anti-angiogenic molecule. In another embodiment, the anti- 
angiogenic molecule is administered in a dose equivalent to that of IMC-lCll, administered 
5 in a dose of from about 5 mg/m^ to about 700 mg/m^ about daily to about every 7 days, more 
preferably a dose of from about 7.5 mg/m^ to about 225 mg/m^, about twice per week. 

The chemotherapeutic agent may be selected from the group consisting of vinca 
alkaloid, camptoihecan, taxane, or platinum analogue, including vincristine, vinblastine, 
vinorelbine, vindesine, paclitaxel, docetaxel, 5 FU, cisplatin, carboplatin, iranotecan, 

10 topotecan or cyclophosphamide. The chemotherapeutic agent is administered in a low-dose 
regimen. Preferably the chemotherapeutic agent is administered at less than about 20% of the 
maximum tolerated dose, more preferably at less than about 15% of the maximum tolerated 
dose, more preferably at less than about 10% of the maximum tolerated dose, more preferably 
at less than about 5% of the maximum tolerated dose, and most preferably at less than about 

15 2% of the maximum tolerated dose. In one embodiment of the invention the 

chemotherapeutic agent is administered at a dose intensity less than about 20% of the dose 
intensity of the chemotherapeutic agent when used in a conventional chem. ^therapeutic 
regimen, pref:ra*' !y at Jose intensity less than about 10% of the dose inte ; of the 
chemotherapeutic agent when used in a conventional chemotherapeutic reeimen, and more 

20 preferably at a dose intensity less than about 5% of the dose intensity of the chemotherapeutic 
agent when used in a conventional chemotherapeutic regimen. 

In one preferred embodiment the vinblastine is administered in a dose from about 0.5 
mg/nr to about 3 mg/m* from about once every 3 days to about once every 7 days. In another 
embodiment, the chemotherapeutic agent is administered in a dosage and frequency that is of 

25 substantially equivalent efficacy to vinblastine in a dose from about 0.5 mg/m^ to about 3 
mg/m- from about once every 3 days to about once every 7 days. Optionally the 
chemotherapeutic agent is administered more frequently than about every three weeks, or 
more frequently than about every seven days. 

The present invention also includes a kit for treating an angiogenic dependent 

30 condition in a mammal comprising the anti-angiogenic molecule and the chemotherapeutic 
agent, which are provided to be administered in an amount and frequency effective, in 
combination, to produce a regression or arrest of the condition while minimizing or 
preventing significant toxicity of the chemotherapeutic agent. 

5 



wo 01/54723 



PCT/USOl/02839 



BRIEF DESCRIPTION OF FIGURES 



Figure 1 is the encoding nucleotide sequence and deduced amino acid sequence of 
and Vl domains of IMC-lCl 1 (c-plCll). 



DETAILED DESCRIPTION OF THE INVENTION 



Throughout this application, various articles and patents, and patent application are 
referenced. Disclosures of all of these publications in their entireties are hereby incorporated 
10 by reference into this application. 

The present invention comprises a method of treating or controlling an angiogenic 
dependent condition in a mammal, comprising administering an anti-angiogenic molecule and 
a chemotherapeutic agent in amounts and frequencies effective to produce, in combination, a 
regression or arrest of the angiogenic dependent condition, while minimizing or preventing 
1 5 significant toxicity. 

The benefits of the combination of an anti-angiogenic molecule and a 
chemotherapeutic agent of the present invention include an improvement in the treatment and 
control of an angiogenic dependant condition with reduced doses of a chemothc .peutic agent 
administered at increased frequency, without significant toxicity. The combination can be 
20 administered for a prolonged period of time, or optionally a shorter duration of treatment may 
be administered due to the increased effectiveness of the combination. Toxicity is reduced or 
eliminated without a loss of effectiveness. The administration of the treatment of the 
invention can overcome the problems of drug resistance that develops with standard 
chemotherapeutic regimens. 

25 

The anti-angiogenic molecule functions to inhibit or prevent angiogenesis, thereby 
treating or controlling the angiogenic dependent condition by inhibiting or blocking 
(antagonizing) the effect of vascular endothelial survival factors. These survival factors are 
receptors or their ligands, upon which vascular endothelium depends, either directly or 
30 indirectly, for growth and/or survival. They play a role in allowing vascular endothelial cells 
to recovery from injury or insult, which, absent the effect of the survival factor would result 
in cell death or apoptosis. Survival factors include vascular endothelial cell growth factors or 
mitogens, as well as those factors which do not appear to have a direct growth-stimulatory 

6 



wo 01/54723 



PCT/USOl/02839 



effect but allow the cells to recover from injury. 

The survival factors tt at are receptors are located on vascular endothelial cells or 
optionally, may be located on other cell types including, but not limited to tumor cells. The 
anti -angiogenic molecule inhibit binding to, and/or activation of, receptors, inhibit their 
5 expression, or inhibit the binding or expression of ligands. 

Examples of survival factors include VEGF receptors, including but not limited to flt- 
1 (VEGFRl), flk-l/KDR (VEGFR2), flt-4 (VEGFR3), their ligands VEGF, VEGF-B, VEGF- 
C, and VEGF-D, integrin aVp3, Tie2/tek, endoglin (GDI 05), neuropilin, thrombospondin 
and their ligands, and PDGFa, PDGFp, aFGF, bFGF, and TGFp, as well as EGF, angiostatin, 
10 and angiopoeitin, vascular permiability factor (VPF), and placenta-like growth factor (PLGF) 
and their receptors. 

Suitable types of anti-angiogenic molecules include, but are not limited to antibody, 
antibody fragment, small molecule or peptide. An antibody can be derived from any 
mammalian species. Optionally, the antibody is of mouse, rat, rabbit, or human origin. 

15 Preferably the antibody is chimeric, more preferably the antibody is humanized, and even 
more preferably the antibody is human. Suitable antibody fragments include, for example. 
Fab fragment. Fab' fragment, F(ab')T fragment, monovalent single chain antibody (scFv), 
and diabodies (DAB ). 

Examples of suitable anti-angiogenic molecules that are antagonists to vascular 

20 endothelium survival factors include, but are not limited to, VEGF receptor antagonist or 
VEGF antagonist, as disclosed in U.S. Patents Nos. 5,840,301, 5,861,499, 5,874,542, 
5,955,31 1, and 5,730,977, which are incorporated in their entirety by reference, aFGF 
receptor antagonist, aFGF antagonist, bFGF receptor antagonist, bFGF antagonist, PDGF 
receptor antagonist, PDGF antagonist, TGFP antagonist, Tie2/tek antagonist (P. Lin et al., 

25 Inhibition of Tumor Angiogenesis Using a Soluble Receptor Establishes a Role for Tie2 in 
Pathologic Vascular Growth. J. Clin. Invest. 100(8) 2072 (1997)), endoglin (GDI 05) 
antagonist, as disclosed in U.S. Patents Nos 5,855,866, and 5,660,827, neuropilin antagonist, 
thrombospondin antagonist, and antagonists to the receptors for PDGFa, PDGpp, aFGF, 
bFGF, or TGpp, as well as antagonists to the receptors for EGF, angiostatin, angiopoeitin, or 

30 VPF (Vascular Permeability Factor) as disclosed in U.S. Patents Nos. 5,036,003 and 

5,659,013. Also encompassed within the scope of the present invention are integrin receptor 
antagonists as disclosed in U.S. Patents Nos. 6,017,926, 6,017,925, 5,981,546, 5,952, 341, 
and 5,919, 792, integrin avP3 antagonists, as disclosed in U.S. Patents Nos. 5,780,426, 



wo 01/54723 



PCT/USOl/02839 



5,773,412, 5,767,071, 5,759,996, 5,753,230, 5,652,1 10, and 5,652,109, antagonists to 
placenta-like growth factor (PLGF) as disclosed in European Patent Application 
EP506477A1, thrombospondin antagonists as disclosed in U.S. Patent Nos. 5,840,692, 
5,770,563, 5,654,277, 5,648,461, 5,506,208, 5,399,667, 5,200,397, 5,192,744, and 5,190,918, 
5 as well as those disclosed in U.S. Patents Nos. 5,965,132, 6,004,555 and 5,877,289, and PCT 
Applications Nos. WO 99/16465, WO 97/05250, WO 98/33917. Also included are molecules 
such as thalidomide, TNP-470, interferon-a (INF-a), and interleukin- 1 2 (IL-12). 

In many cases, the expression of a receptor and/or ligand is upregulated in an region 
of angiogenesis. However, although located in an area of abnormal cells responsible for the 

10 specific disease, exposed to high levels of ligand, and having upregulated receptors, the cells 
of the vascular endothelium are largely normal and responsive to normal regulatory 
mechanisms. Because the receptors exist on essentially normal endothelial cells, their 
behavior is less likely to escape normal regulatory control. An advantage to blocking a 
receptor, rather than its ligand, is that fewer anti-angiogenic molecules may be needed to 

1 5 achieve such inhibition, as levels of receptor expression may be more constant than those of 
the environmentally induced ligand. Although there are advantages to targeting receptors, it is 
also possible, and within the scope of the present invention, to inhibit angiogenesis by 
targeting the ligand for the receptor, either alone or in combination with blockade of the 
receptor. Optionally, antagonism of the receptor is combined with antagonism of the ligand in 

20 order to achieve even more efficient inhibition of angiogenesis. 

A preferred embodiment of the invention is the combination of a chemotherapeutic 
agent and a VEGF receptor antagonist. It has been shown that a major function of VEGF is to 
promote the survival of endothelial cells comprising newly formed vessels (L. E. Benjamin, 
et AL, Selective Ablation of Immature Blood Vessels in Established Human Tumors Follows 

25 Vascular Endothelial Growth Factor Withdrawal. J,Clin. Invest. 103:159-165 (1999), T. Alon, 
et aL, Vascular Endothelial Growth Factor Acts as a Survival Factor for Newly Formed 
Retinal Vessels and Has Implications for Retinopathy of Prematurity. Nature Med. 1 : 1024- 
1028 (1995), R.K. Jain, et al.. Endothelial Cell Death, Angiogenesis, and Microvascular 
Function after Castration in an Androgen-Dependent Tumor: Role of Vascular Endothelial 

30 Growth Factor. Proc. Natl Acad. Scl U.S.A. 95:10820-10825 (1998)) Hence, the ability of 
such cells to cope with the damage inflicted by continuous or frequent exposure to a 
chemotherapeutic drug is selectively and significantly impaired when they are exposed to a 
VEGF receptor antagonist, ( M. J. Prewett, et al., Antivascular Endothelial Growth Factor 

8 



wo 01/54723 



PCT/USOl/02839 



Receptor (Fetal Liver Kinase 1) Monoclonal Antibody Inhibits Tumor Angiogenesis and 
Growth of Several Mouse and Human Tumors. Cancer Res 59:5209-5218. (1999); T. A. 
Fong, et al., SU5416 Is a Potent and Selective Inhibitor of the Vascular Endothelial Growth 
Factor Receptor (Flk-l/kdr) That Inhibits Tyrosine Kinase Catalysis, Tumor Vascularization, 
5 and Growth of Multiple Tumor Types. Cancer Res 59:99-106 (1999); N. Ferrara, et al.. 

Clinical Applications of Angiogenic Growth Factors and Their Inhibitors. Nat.Med. 5:1359- 
1364. (1999)). It is believed that the combination of continuous or frequent chemotherapy 
with, for example, interruption of the cell rescue mechanisms provided by activation of the 
VEGF receptor plays a role in inducing vascular endothelial cell apoptosis. 

10 In a preferred embodiment of the invention, the anti-angiogenic molecule is an 

antagonist to VEGF or the VEGF receptor. While the expression of the VEGF receptor and 
ligand is low in normal endothelial cells that are not in or near a region of angiogenesis, 
VEGF receptors present on tumor infiltrating vascular endothelial cells are upregulated, as is 
the expression of the VEGF ligand by tumor cells. Blocking the interaction between VEGF 

15 and its receptors can inhibit angiogenesis, and thereby tumor growth, while not significantly 
effecting normal endothelial cells at other sites, where vascular endothelial cell receptors have 
not been upregulated. In one embodiment of the present invention, antagonism of the VEGF 
receptor is combined with antagonism of the VEGF ligand in order to achieve even more 
efficient inhibition of angiogenesis. In other embodiments of the invention antagonists to one 

20 or more than one of the VEGF receoptors or ligands are administered. VEGF (or VEGF-A) is 
the ligand for VEGFRl and VEGFR2, VEGF-B is the ligand for VEGFR2, VEGF-C is the 
ligand for VEGFR3, VEGFR4, and possibly VEGFR2, and VEGF-D is the Ugand for 
VEGFR2 and VEGFR3. Optionally, the effect of more than one form of VEGF is inhibited. 
An example of an antagonist to a VEGF receptor (flk-1) is the antibodies DClOl, 

25 described in the Examples. Another is A.4.6. 1 and its chimeric and humanized form as 
disclosed in L. G. Presta, Humanization of an Anti-vascular Endothelial Growth Factor 
Monoclonal Antibody for the Therapy of Solid Tumors and Other Disorders. Cancer 
Research^ 57, 4593-4599 (1997), which is hereby incorporated by reference. A preferred 
VEGF antagonist is the mouse-human chimeric antibody IMC-lCl 1 which is a KDR 

30 antagonist, and is disclosed in U.S. Application 09/240,736, which is hereby incorporated by 
reference. The encoding nucleotide sequences and deduced amino acid sequences of the Vh 
and Vl domains are shown in Figure 1 . 

The chemotherapeutic agent of the present invention functions, in combination with 

9 



wo 01/54723 



PCT/USOl/02839 



the anti-angiogenic molecule, to cause a cytotoxic effect on the vascular endothelial cells 
involved in angiogenesis. A number of chemotherapeutic agents have been identified as 
having anti-angiogenic activity and are suitable for use in the practice of the present 
invention. Examples include, but are not limited to, taxanes, including but not limited to 
5 paclitaxel and docetaxel, camptothecin analogues, including but not limited to iranotecan and 
topotecan, platinum analogues including but not limited to cisplatin and carboplatin, 5FU, 
and vinca alkaloids, including but not limited to vinblastine, vincristine, vindesine and 
vinorelbine. 

The present invention provides a low dose application of a chemotherapeutic agent 

10 administered in combination with an anti-angiogenic molecule in an amount and frequency 
that, in combination, provides effective therapy without significant side-effects. Effective 
therapy is therapy that provides regression or arrest of the angiogenic dependant condition. 
An effective amount of anti-angiogenic molecule and chemotherapeutic agent is an amount of 
each, that in combination controls (causes regression or arrest) the condition being treated 

15 without producing significant chemotherapy induced toxicity. The meaning of significant 
toxicity is well known to one of ordinary skill in the art, and includes toxicities that 
cumulatively or acutely effect a patient's quality of life and/or limit the amount of 
chemotherapeutic agent than can be admiivistered. 

Examples of chemotherapy induced toxicity that can be minimized or prevented by 

20 the present invention include, but are not limited to. myelosuppression, neurotoxicity, 

cardiotoxicity, alopecia, nausea and vomiting, nephrotoxicity, and gastrointestinal toxicity. 
The low dose administration of a chemotherapeutic agent without significant toxicity permits 
prolonged treatment if desired. Additionally, the low dose manner of chemotherapy 
administration in the present invention can overcome the problem of the development of 

25 chemotherapeutic drug resistance by the patient's tumor cells that occurs with current 

chemotherapeutic regimens which consist of higher doses of drug administered intermittently 
with longer time intervals between treatment. The present invention delays, reduces, or even 
circumvents the problem of acquired drug resistance by targeting the genetically stable 
endothelial cells of newly formed tumor blood vessels, rather than genetically unstable tumor 

30 cells which are prone to mutate and develop resistance. Encompassed within the scope of the 
present invention is the administration of amounts of chemotherapy that are insufficient to 
have a cytotoxic effect on tumor cells yet have anti-angiogenic properties as a result of the 
drug's effect on vascular endothelial cells. 

10 



wo 01/54723 



PCT/USOl/02839 



The low-dose administi ation of chemotherapeutic agents, to achieve therapeutic 
effects without significant toxicity (side effects) is readily possible by the practice of the 
present invention. Applying standard methods of defining optimal dosage levels and 
schedules to the teachings of the present invention, one of ordinary skill in the art readily can 
5 determine a more or most desirable low-dose regimen for a selected chemotherapeutic agent 
when used in combination with an anti-angiogenic molecule, as detailed in the present 
application. A low-dose regimen will administer the chemotherapy at frequent intervals or 
continually, at less than about 50% of the maximum tolerated dose (MTD), more preferably 
less than about 45% of the MTD, more preferably less than about 40% of the MTD, more 

10 preferably less than about 35% of the MTD, more preferably less than about 30% of the 
MTD, more preferably at less than about 25% of the MTD, more preferably at less than 
about 20% of the MTD, more preferably at less than about 10% of the MTD more 
preferably less than about 5% of the MTD, and most preferably at less than about 2% of the 
MTD, although the preferred dose depends on the particular chemotherapy. In any event, the 

15 preferred dose will be a dose effective to inhibit or prevent progression of the angiogenic 

dependent condition, when administered in combination with the anti-angiogenic molecule of 
the present invention, while minimizing or preventing the development of significant 
chemotherapy related toxicity. Optionally :1 . dose of chemotherapy will be effective to 
inhibit or prevent progression of the angiogenic dependent condition even when administered 

20 alone, although it is not intended that it be administered in this manner. Optionally the dose 
of chemotherapy will be one which is sufficiently low that it does not exert a direct cytotoxic 
effect on tumor cells, yet has an antitumor effect mediated by its anti-angiogenic properties. 

Optionally, the low-dose regimen of the present invention will administer the 
chemotherapeutic agent at a dose intensity of less than about 20% of the dose intensity used 

25 when the chemotherapeutic agent is administered as part of a conventional chemotherapeutic 
regimen (i.e. administered at connventional dosages and frequencies without an anti- 
angiogenic molecule or other treatment modality) used to treat a particular neoplasm. The 
dose intensity of the chemotherapeutic agent used in a conventional regimen can be readily 
determined by one of ordinary skill in the art. By way of example, various regimens are 

30 disclosed in V.T. Devita et al., Cancer: Principles & Practice of Oncology^ 5th edition, 
Lippencott Williams and Wilkins. (1997) More preferably the present invention will 
administer the chemotherapeutic agent at a dose intensity of less than about 1 0% of that used 
when the chemotherapeutic agent is administered as part of a conventional chemotherapeutic 

11 



wo 01/54723 



PCT/USOl/02839 



regimen used to treat a particular neoplasm, and most preferably at a dose intensity of less 
than about 5% of that used when the chernotherapeutic agent is administered as part of a 
conventional chemotherapeutic regimen used to treat a particular neoplasm. 

In the prior art, chemotherapy is usually given intermittently, commonly in the form 
5 of a bolus infusion or an infusion lasting from about 20 minutes to about three hours, at about 
the maximum tolerated dose (MTD) with long rest periods (e.g., 3 weeks) between successive 
drug exposures. It has been suggested that these rest periods provide the endothelial cell 
compartment of a tumor an opportunity to repair some of the damage inflicted by the 
chemotherapy (T. Browder, et al., Antiangiogenic Scheduling of Chemotherapy Improves 

1 0 Efficacy Against Experimental Drug- Resistant Cancer. Cancer Res. (In press) 1 999). 

Administering lower doses of a chemotherapeutic drug more frequently such as weekly, more 
preferably several times a week or continuously, enables circumvention of many problems 
associated with standard chemotherapeutic doses. This anti-angiogenic scheduling of 
chemotherapy optimizes anti-vascular effects. An added benefit is that administration of 

15 chemotherapy in this manner can result in the increased sensitivity of the tumor cells to 

chemotherapy. For example, a sub-line of the Lewis Lung Carcinoma, previously selected in 
vivo for acquired resistance to the MTD of cyclophosphamide, is rendered sensitive again to 
the drug in vivo by employing continuous \o\\' dose ir ;^apy of the same drug. The inclusion 
of the anti-angiogenic molecule of the present invention provides substantial and 

20 unexpectedly better results. 

The invention provides low-dose administration of chemotherapy administered at 
short intervals, preferably more frequently than every three weeks, more preferably more 
frequently than weekly. Most preferably it is administered from about every 4 to about every 
6 hours, to about daily to weekly. Optionally it is administered continuously. The preferred 

25 time interval between administration of successive doses of chemotherapeutic agent is that 
amount of time that is of sufficiently short duration that the blood levels of the 
chemotherapeutic agent (or its active metabolite) will remain at about a concentration 
sufficient to exert an anti-angiogenic effect for substantially the duration of treatment. 
Preferably, such a blood level will be maintained for at least about 20% of the time between 

30 doses, more preferably for at least about 30% of the time between doses more preferably for 
at least about 50% of the time between doses, most preferably for at least about 70% of the 
time between doses. Therapy is continued for a period of time from about 10 days to about 6 
months, or as determined by one of skill in the art. Optionally, treatment will continue 

12 



wo 01/54723 



PCT/USOl/02839 



chronically for a period longer than six months for as long as is needed. The present invention 
reduces host toxicity, allows for longer term administration of the chemotherapeutic agent in 
diseases or pathological conditions requiring it, and does not sacrifice, and perhaps even 
improves, anti-tumor efficacy. Optionally, increased efficacy will permit the use of shorter 
5 durations of therapy for selected angiogenic dependent conditions. 

The anti-angiogenic molecule is preferably administered in dosages and dose 
fi-equencies sufficient to substantially saturate the selected target receptor or ligand. 
Substantial saturation is saturation of at least about 50% of targeted receptors. A more 
preferred level of saturation is at least about 80%, and a most preferred level of saturation is 
10 about 100%. Optionally, the anti-angiogenic molecule is administered at a dose and 

frequency sufficiently short to maintain a blood level sufficient to saturate the targeted 
survival factor for at least about 50% of the time between doses, more preferably at least 
about 70% of the time and most preferably at least about 90% of the time interval between 
doses. Using the concentrations required to achieve receptor saturation or ligand 
15 neutralization in vitro, and by analysis of serum concentrations of anti-angiogenic molecule 
in vivo, both the appropriate dose and schedule can be determined readily by one of skill in 
the art. 

A preferred embodiment of the present invention c omprises the administration of the 
antibody IMC-lCl 1, a KDR receptor antagonist with a c lemotherapeutic agent. A preferred 

20 dose of IMC-lCl 1, is an amount that is sufficient to adequately saturate the targeted 

receptors or ligand. In in vitro experiments, 50% saturation of VEGF receptors was obtained 
as an IMC-lCl 1 concentration of 0.2 |ig/ml, and 100% at a concentration of 3 (ig/ml. A 
preferred level of saturation is about at least 50%, a more preferred level is about at least 
80%, and a most preferred level is about 1 00% saturation. For therapy, a preferred dose 

25 regimen of IMC-lCl 1 is from about 5 mg/m- to about 700 mg/m-, more preferably from 
about 7.5 mg/m' to about 225 mg/m-, administered about twice per week. 

Another preferred embodiment of the invention combines IMC-lCl 1 in the doses 
described above with vinblastine, administered in a low dose regimen, at a dose from about 
0.5 mg /nr to about 3 mg/m- from about every 3 days to about every 7 days. Optionally, a 

30 suitable chemotherapeutic agent other than vinblastine is administered in a dosage and 

frequency that is of substantially equivalent efficacy to vinblastine (in the combination) at a 
dose from about 0.5 mg /m- to about 3 mg/m- from about every 3 days to about every 7 days. 
In other embodiments of the present invention, doses of an anti-angiogenic molecule 

13 



wo 01/54723 



PCT/USOl/02839 



in amounts and dosing frequencies sufficient to provide levels of receptor or ligand saturation 
equivalent to that of IMC-lCl 1 in the doses about are combined with a chemotherapeutic 
agent in a dose and frequency equivalent to that of vinblastine above, and therapy is carried 
out for as long as is needed. An equivalent dose is one that, in the combination, is 
5 substantially as effective in arresting or inhibiting the angiogenic dependent condition, while 
being substantially as effective in minimizing or preventing significant chemotherapy 
induced toxicity. In one preferred embodiment of the present invention, an equivalent dose of 
another chemotherapeutic agent is determined using data derived from an animal model, an 
example of which is included herein, utilizing a chemotherapy-resistant cell line so that any 

10 observed antitumor effect is due to an effect on the vascular endothelium. A preferred dose of 
vinblastine in a mouse is from about 1 mg/m^ to about 2 mg/m^ more preferably about 1.5 
mg/m^ administered every three days. The MTD of this drug in mice is approximately 4-5 
times that of a human, and a preferred dose is 1/16 - 1/20 of the MTD in mice. A preferred 
dose of DC 101 in a mouse is about 800 \ig administered intraperitoneally every three days. 

15 The use of DC 101 and vinblastine showed a therapeutic effect upon neuroblastoma cell lines 
grown as xenografts in SCID mice. (L. Witte, L, et al.. Monoclonal antibodies targeting the 
VEGF receptor-2 (flkl/KDR) as an anti-angiogenic therapeutic slvaXegy. Cancer Metastasis 
Rev. 1 7: 1 55-161. (1998); Prewitt, 1999). In yet another preferred embodiment of the present 
invention, low-dose vinblastine is administered every 3 days in combination with IMC- 1 CI 1 

20 (plCll). 

The anti-angiogenic molecule and chemotherapeutic agent of the present invention are 
administered together or separately. Routes of administration include but are not limited to 
oral, sublingual, and parenteral, including intravenously, subcutaneously, transcutaneously, 
intraperitoneally, intrapleurally, and intrathecally. Optionally the molecule and agent are 

25 formulated into a pharmaceutical preparation for administration via the desired route. The 
agent and molecule are administered via the same route or via different routes 

In one aspect of the present invention, there is provided a kit comprising an anti- 
angiogenic molecule and a chemotherapeutic agent to be administered to a mammal in an 
amount effective to produce a regression or arrest of an angiogenic dependent condition while 

30 minimizing or preventing significant toxicity of the chemotherapeutic agent. Such a kit 

optionally comprises an anti-angiogenic molecule and a chemotherapeutic agent in one or 
more than one containers for administration at about the same time points or at different 
times. Optionally, the anti-angiogenic molecule is administered intermittently and the 

14 



wo 01/54723 



PCT/USOl/02839 



chemotherapeutic agent is adnr inistered continuously or in a manner that permits the 
maintenance of a suitable bloo J concentration. It is an aspect of the present invention that 
such treatment optionally is administered for a prolonged period or chronically, without 
substantial chemotherapy induced toxicity. Routes of administration include but are not 
5 limited to oral and parenteral, including but not limited to intravenous, subcutaneous, 

percutaneous, intrathecal and intraperitoneal. Patients that may be treated with the methods 
and compositions of the present invention include any patients with an angiogenic dependent 
disease. 

The angiogenic dependent diseases encompassed by the scope of the present invention 

10 include, but are not limited to neoplasms, collegen vascular diseases or autoimmune diseases. 
All neoplasms are suitable for treatment with the present invention, however preferred 
neoplasms are solid tumors. More preferred are breast carcinoma, lung carcinoma, prostate 
carcinoma, colon carcinoma, prostate carcinoma, ovarian carcinoma, neuroblastoma, central 
nervous system tumor, neuroblastoma, glioblastoma multiforme or melanoma, and a 

15 preferred mammal to receive treatment is a human. 

Antibodies used in this invention may be produced in a prokaryotic or eukaryotic cell. 
Techniques for the creation of and production of such antibodies, or portions thereof are well 
know in the field and are within the knowledge of one of ordinary skill in the art. Techniques 
used for preparation of monoclonal antibodies, include but are not limited to, the hybridoma 

20 technique (Kohler & Milstein, Nature. 256:495-497 (1975)), the trioma technique, the human 
B-cell hybridoma technique (Kozbor et al., Immunology Today 4:72, (1983)), and the EBV- 
hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1985, In 
Monoclonal Antibodies and Cancer Therapy^ Alan R. Liss, Inc., pp. 77-96). 

DNA encoding chimerized antibodies may be prepared by recombining DNA 

25 substantially or exclusively encoding human constant regions and DNA encoding variable 
regions derived substantially or exclusively from the sequence of the variable region of a 
mammal other than a human. DNA encoding humanized antibodies may be prepared by 
recombining DNA encoding constant regions and variable regions, other than the CDRs, 
derived substantially or exclusively from the corresponding human antibody regions and 

30 DNA encoding CDRs derived substantially or exclusively from a mammal other than a 
human. 

The DNA deletions and recombinations of the present invention may be carried out by 
known methods, such as those described in PCT applications WO 93/21319, WO 89/09622, 



wo 01/54723 



PCT/USOl/02839 



European Patent applications 239,400, 338,745 and 332,424 and/or other standard 
recombinant DNA techniques. Conventional methods, such as those employed in the 
construction of vectors and piasmids, the insertion of genes encoding polypeptides into such 
vectors and piasmids, or the introduction of piasmids into host cells, are well known to those 
5 of ordinary skill in the art and are described in numerous publications including Sambrook, J., 
Fritsch, E.F. and Maniatis, T. (1989) Molecular Cloning: A Laboratory Manual, 2nd edition. 
Cold Spring Harbor Laboratory Press, and in Ausubel et al. (Eds) Current Protocols in 
Molecular Biology, Green Publishing Associates/ Wiley-Interscience, New York (1990). 

The invention also includes functional equivalents of the antibodies described in this 
10 specification. Functional equivalents have binding characteristics comparable to those of the 
antibodies, and include, for example, chimerized, humanized and single chain antibodies as 
well as fragments thereof. Methods of producing such functional equivalents are disclosed in 
PCX Application No. WO 93/21319, European Patent Application No. EPO 239,400; PCT 
Application WO 89/09622; European Patent Application No. EP338,745; and European 
1 5 Patent Application EPO 332,424. 

The present invention also includes chimeric, single chain, and humanized antibodies, 
as well as Fab fragments, or the product of an Fab expression library. The antibodies of the 
invention can prepared by conventional methods which are well know in the art. 

Techniques described for the production of single chain antibodies (U.S. Pat. No. 
20 4,946,778, incorporated herein by reference) are adapted to produce single chain antibodies to 
immunogenic polypeptide products of this invention. 

According to another embodiment of the invention, the antibodies of the invention can 
be prepared by recombinant DNA techniques by cloning and expressing all or part of a 
known antibody. Using such techniques, which are known in the art, a chimeric or 
25 humanized version of non-human antibodies can be prepared- For example, a chimeric 

or humanized version of monoclonal antibody can be readily prepared by cloning the gene 
encoding this antibody in to an appropriate expression vector. Useful in this regard are the 
nucleic acids which encodes an amino acid sequence wherein the amino acid sequence 
comprises the variable region, hypervariable region, or both of a monoclonal antibody that 
30 specifically binds to a vascular endothelial survival factor. 

More particularly, the present invention also includes recombinant constructs 
comprising one or more of the sequences as broadly described above. The constructs 
comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention 

16 



wo 01/54723 



PCTAJSOl/02839 



has been inserted, in a forward or reverse orientation. In a preferred embodiment of this 
embodiment, the construct further comprises regulatory sequences, including, for example, a 
promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters 
are known to those of skill in the art, and are commercially available. The following vectors 
5 are provided by way of example. Bacterial: pQE70, pQE60, pQE-9 (Qiagen), pbs, pDlO, 
phagescript, psiX174, pbluescript SK, pbsks, pNH8A, pNH16a, pNHlSA, pNH46A 
(Stratagene); ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia). Eukaryotic: 
pWLNEO, pSVZCAT, pOG44, pXTL pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL 
(Pharmacia). However, any other plasmid or vector may be used as long as they are replicable 

1 0 and viable in the host. 

The constructs in host cells are used in a conventional manner to produce the gene 
product encoded by the recombinant sequence. Appropriate cloning and expression vectors 
for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al.. Molecular 
Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989). 

1 5 According to another aspect of the invention, transgenic mammals are provided that 

express humanized antibodies to immunogenic products of this invention. Novel transgenic 
mammalian hosts, other than primates, particularly other than human, are provided, where the 
host is capable of mounting an immune response to an immunogen, where the response 
produces antibodies having primate, particularly human, constant and/or variable regions or 

20 such other effector peptide sequences of interest. 

The hosts are characterized by being capable of producing xenogenic or modified 
antibodies as a result of substitution and/or inactivation of the endogenous immunoglobulin 
subunit encoding loci. The modifications retain at least a portion of the constant region which 
provides for assembly of the variable region binding site bonded at the C-terminus to a 

25 functional peptide. The functional peptide takes many forms or conformations and serves, for 
example, as an enzyme, growth factor, binding protein, ligand, cytokine, effector protein, 
chelating proteins, etc. The antibodies are any isotype, i.e., IgA, IgD, IgE, IgG, IgM or 
subtypes within the isotype. 

Transgenic hosts include murine, lagomorpha, ovine, porcine, equine, canine, feline, 

30 and the like. For the most part, mice have been used for the production of B-lymphocytes. It 

should be understood that other animals may be readily substituted for the mice, following 

the same procedures. 

Humanized and chimeric antibodies are prepared according to the following 

17 



wo 01/54723 



PCT/USOl/02839 



strategies. In one strategy, the human heavy and light chain immunoglobulin gene complexes 
are introduced into the mouse germ line and in a separate step the corresponding mouse genes 
are rendered non-functional. Polynucleotides encoding human heavy and light chain are 
reconstructed in an appropriate eukaryotic or prokaryotic microorganism and the resulting 
5 polynucleotide fragments are then introduced into pronuclei of fertilized mouse oocytes or 

embryonic stem cells. Inactivation of the endogenous mouse immunoglobulin loci is achieved 
by targeted disruption of the appropriate loci by homologous recombination in mouse 
embryonic stem cells. In each case chimeric animals are generated which are derived in part 
from the modified embryonic stem cells and are capable of transmitting the genetic 
10 modifications through the germ line. The mating of mouse having a human immunoglobulin 
loci to mouse having an inactivated immunoglobulin loci yields animals that produce purely 
human antibody. 

In another strategy, fragments of the human heavy and light chain immunoglobulin 
loci are used to directly replace the corresponding mouse loci by homologous recombination 

15 in mouse embryonic stem cells. This is followed by the generation of chimeric transgenic 
animals. The resulting human antibodies are isolated, for example, from other proteins by 
using an affinity column, having an Fc binding moiety, such as protein A, or the like. 

The organi::ai:on, relative location of exons encoding individual domains, and 
location of splice sites and transcriptional elements in a number of animals are known by 

20 those of ordinary skill in the art. In human, for example, the immunoglobulin heavy chain 
locus is located on chromosome 14. In the 5'-3' direction of transcription, the locus 
comprises a large cluster of variable region genes (VH), the diversity (D) region genes, 
followed by the joining (JH) region genes and the constant (CH) gene cluster. The size of the 
locus is estimated to be about 2,500 kilobases (kb). During B-cell development, 

25 discontinuous gene segments from the germ line Ig H locus are juxtaposed by means of a 
physical rearrangement of the DNA. 

Production of a functional heavy chain immunoglobuline polypeptide requires three 
discontinuous DNA segments, from the VH, D, and JH regions, to be joined in a specific 
sequential fashion generating the functional units. Once these units are formed specific heavy 

30 chains are produced following transcription of the immunoglobuline locus. There are two 
loci for immunoglobuline light (Ig L)chains, the kappa locus on human chromosome 2 and 
the lambda locus on human chromosome 22. The structure of the Ig L loci is similar to that 
of the Ig H locus, except that the D region is not present. 

18 



wo 01/54723 



PCT/USOl/02839 



The entire V region, or various fragments of the V region is used to produce a broad 
spectrum of high affinity antibodies. For example, a subset of the known V region genes of 
the human heavy and light chain Ig loci (Herman et al., EMBO J. 7: 727-738 (1988)) is used 
to produce transgenic hosts, which transgeijic host are capable of mounting a strong immune 
5 response and provide high affinity antibodies. 

Antibodies or antibody analog producing B-cells from the transgenic host are used, 
for example, for fusion to a mouse myeloid cell to produce hybridomas or immortalized by 
other conventional process, i.e., transfection with oncogenes. These inmiortalized cells are 
then grown, for example, in continuous culture or introduced into the peritoneum of a 
10 compatible host for production of ascites. 

As discussed above, present invention also provides for the production of polyclonal 
human anti-serum or human monoclonal antibodies or antibody analogs provided they retain 
the activities of the antibodies of the invention. Epitope binding component of the present 
invention refers to proteins consisting of one or more polypeptides substantially encoded by 
15 genes of the immunoglobulin superfamily (i.e.. The Immunoglobulin Gene Superfamily, 
Williams & Barclay In: Immunoglobulin Genes, Honjo, Alt, and Rabbitts, eds., (1989) 
incorporated herein by reference). For example, an epitope binding component comprises 
part or all of a heavy chain, part or all of a light chain, or both. However, an cpitupe binding 
component must contain a sufficient portion of an immunoglobulin superfamily gene product 
20 to retain the ability to bind to a specific target, or epitope. 

Included within the scope of this invention is bispecific antibodies that are formed by 
joining two epitope binding components that have different binding specificities. 

In general, modifications of the genes encoding the desired epitope binding 
components are readily accomplished by a variety of well-known techniques, such as site- 
25 directed mutagenesis (see, Gillman & Smith, Gene 8:81-97 (1979) and Roberts, et. al.. Nature 
328:731-734 (1987), both of which are incorporated herein by reference). 

In preferred embodiments of the invention, the epitope binding component of the 
antibody of this invention is encoded by immunoglobulin genes that are "chimeric" or 
"humanized" (see, generally. Queen (1991) Nature 351:501, which is incorporated herein by 
30 reference). Once expressed, VE-cadherin antibodies, epitope binding components, their 

dimers, or individual light and heavy chains are purified according to standard procedures of 
the art, for example, ammonium sulfate precipitation, fraction column chromatography, gel 
electrophoresis and the like (see, generally. Scopes, Protein Purification, Springer-Verlag, 

19 



wo 01/54723 



PCT/USOl/02839 



N.Y. (1982)). Once purified, partially or to homogeneity as desired, the antibodies and 
fragments thereof are then used ,for example, therapeutically, diagnostically, in drug 
screening techniques, or in developing and performing assay procedures, such as 
immunofluorescent staining, and the like. 
5 The examples which follow are set forth to aid in understanding the invention, but are 

not intended to, and should not be construed as, limiting the scope of the invention in any 
manner. 



20 



wo 01/54723 



PCT/USOl/02839 



Examples 

Cells and culture conditions: Neuroblastoma cell lines SK-N-MC, SK-N-AS were 
obtained from American Type Culture Collection (ATCC) and expanded as a monolayer 
culture by serial passage on tissue culture plates (Nunc, Denmark) in DMEM, 5% fetal 
bovine serum (Gibco, Grand Island, NY, USA). Human umbilical vein endothelial cells 
(HUVEC) (Clonetics, San Diego, CA) were expanded on 1% gelatin-coated tissue culture 
plates in MCDBI31 culture medium (JRH Biosciences, Lenexa, KS, USA) supplemented 
with 5 ng/ml bFGF (R&D, Minneapolis, MN), 10 units/ml heparin (Wyeth-Ayerst Canada), 
10 ng/ml EGF (UBI, Lake Placid, NY) and 10% fetal bovine serum. 

In vitro determination of drug sensitivity; Three thousand cells in 200 jil growth 
media per well were plated in 96-well flat bottom tissue cuhure plates (Nunc, Denmark) and 
incubated at 37 °C, 5% CO2 for 24 hours prior to initiation of treatment. The cells were then 
washed with PBS and treated with 1-500 ng/ml vinblastine sulphate (Calbiochem, La Jolla, 
Ca) for 24 hours, in groups of eight wells per dose. The cells were then pulsed for 6 hrs with 
2 |iCi/well of methyl-^H-thymidine (Amersham Life Science, Buckinghamshire, England). 
The plates were frozen, thawed and the DNA harvested onto a filtermat using a Titertek Cell 
Harvester. Incorporated radioactivity was measured on Wallac 1205 BetaPlate Scintillation 
Counter (Wallac Oy, Finland) and proliferation was expressed as a percentage of ^H- 
thymidine in treated cells vs. that in controls. 

In vivo tumor growth assessment: SK-N-MC, cells were harvested using 1% Trypsin 
EDTA (GibcoBRL, Gaithesburg, MD), and single cell suspension of 2 x 10^ cells in 0.2 ml of 
growth media was injected subcutaneousiy into the flanks of 4-6 week old CB-1 7 SCID mice 
(Charles River, St-Constant, Quebec). Approximately 3 weeks later, most tumors had grown 
to — 0.75 cm^, and mice were randomized into groups of 5 animals. Two independent 
experiments were performed, each totaling 20 animals in 4 groups. The treatment was as 
follows: 

Group I (Control) - 0.4 ml of PBS (DC 101 vehicle) i.p. every three days and 0.15 ml 
injectable saline (vinblastine vehicle) i.p. every three days. 

Group II - 0.4 ml of 2 mg/ml DC 101 antibody (800 |ag/mouse) (24) every three days 
and 0.15 ml of injectable saline i.p. every three days 

21 



wo 01/54723 



PCT/USOl/02839 



Group III - vinblastine sulfate 0.75 mg/m^ i.p. bolus at the start of therapy, followed 
by 1 mg/mVday via subcutaneous Alzet osmotic pumps (Alza Corp,Palo Alto, CA) for 3 
weeks, followed by maintenance therapy with 0. 1 5 ml of 0.067 mg/ml vinblastine sulfate (1 .5 
mg/m^) i.p. every three days, and 0.4 ml of PBS i.p. every three days 
5 Group IV - combination of DC 1 01 and vinblastine at doses identical to the single 

agent groups. 

The body weight, tumor size and general clinical status of the animals were recorded 
every 2-3 days. Perpendicular tumor diameters were measured using a vernier scale caliper 
and tumor volume was estimated using the formula for ellipsoid: (width-^ x length)/2. Growth 
10 curves were statistically analyzed using repeated measures ANOVA. All animal care was in 
accordance with institutional guidelines. As required by institutional guidelines, the mice 
were sacrificed when tumor size reached 1 .5 cm^ or 7.5-10% of their body weight. 

Histology: All tumors were excised, fixed in 10% (v/v) formalin and processed for 
15 immunohistochemical analysis. To obtain adequate tissue for the combination treatment 

group, two mice were sacrificed at 7.5 weeks of treatment. Paraffin blocks were cut to 5 \im 
sections and stained with haematoxylin/eosin for morphology evaluation and with Apoptosis 
Detection System (Promega, Madison, Wisconsin) for assessment of programmed cell death. 

20 Relative tumor vascularity assessed by an FITC-Dextran perfusion assay: The 

method was designed to assess the relative functionality of the tumor vasculature. 2x10^ SK- 
N-AS neuroblastoma cells were injected into the flanks of CB-17 SCID mice. Tumors were 
allowed to grow to approximately 0.75 cm^ at which point tumor bearing mice were then 
treated with 1 mg/m~ vinblastine i.p. every three days, 800 ^ig DC 101 i.p. every three days, 

25 combination of the two agents or saline as a control. At 14 days, when divergence in tumor 
growth between the treatment groups was clearly evident, 0.2 ml of 25 mg/ml FITC-Dextran 
in PBS (^igma, St. Louis, MO) was injected systemically into the lateral tail vein of each 
mouse and allowed to circulate for 20-30 minutes. Mice were then sacrificed by cervical 
dislocation and blood samples were collected into heparinized tubes by cardiac puncture for 

30 assessment of systemic fluorescein levels. Tumors were resected from the surrounding 
connective tissue being careful to avoid spillage of intra-vascular contents, weighed and 
placed into tubes containing 1:10 dispase (Collaborative Research, Two Oaks, Bedford, MA). 
To normalize for dilution caused by the difference in tumor sizes, 1 ml of 1 : 10 dispase was 

22 



wo 01/54723 



PCT/USO 1/02839 



added per 0.5 g of tissue. Tumors were incubated in a dark 37 °C shaker ove. night. The tissue 
was homogenized, centrifuged at 5000 rpm for 1 0 minutes, and the supernatant was collected 
and stored in the dark until further analysis. Blood samples were centrifuged immediately 
following collection, plasma separated and protected fi*om light at 4°C until analysis. 
Fluorescence readings were obtained on a FL600 Fluorescence Plate Reader (Bio-tek 
Instruments Inc., Winooski, Vermont, USA), from a standard curve created by serial dilution 
of the FITC-dextran used for injection. The ratio of tumor fluorescence: plasma fluorescence 
was assumed to be reflective of the degree of tumor perfusion. 

In vivo angiogenesis assessment by the Matrigel plug assay (5,25): Matrigel 
(Collaborative Biomedical Products, Bedford, MA) stored at -20 °C, was thawed at 4°C 
overnight and mixed with 500 ng/ml bFGF. 0.5 ml of this mixture was then injected 
subcutaneously into the shaved flanks of twenty 6-8 week old female Balb/cJ mice (Jackson 
Labs, Bar Harbor, Maine). Five mice, used as negative controls, were injected with Matrigel 
alone. Three days later, treatment mice were randomized into four groups as follows: 

Group 1 - saline i.p.. 

Group II - 800 jag DCIOl i.p. 

Group III - 1 mg/m- virHlastine i.p. 

Group IV - combination therapy. 
All 25 mice were treated on day 4 and 7 and sacrificed on day 10. Blood samples were 
collected into heparinized tubes by cardiac puncture, centrifuged immediately following their 
collection, plasma separated and protected from light at 4°C. The Matrigel plugs were 
resected from surrounding connective tissues, placed into tubes containing 1 ml of 1 : 1 0 
dispase and incubated in the dark in a 37 °C shaker overnight. The following day, the plugs 
were homogenized, centrifuged at 5000 rpm for 1 0 minutes and supernatant saved in the dark 
for analysis of fluorescence. Fluorescence readings were obtained on FL600 Fluorescence 
Plate Reader using a standard curve created by serial dilution of FITC-dextran used for 
injection. Angiogenic response was expressed as a ratio of Matrigel plug fluorescence: 
plasma fluorescence. 

In vitro determination of differential drug sensitivity: Prior to undertaking our \n vivo 
experiments we established a dose of vinblastine, at which significant toxicity of endothelial, 
but not tumor, cells was observed. To do so, we optimized growth conditions to achieve 

23 



wo 01/54723 



PCT/USOl/02839 



comparable levels of mitotic activity in tw^o human neuroblastoma cell lines (SK-NM-C and 
SK-N-AS) and HUVEC. All three cell lines were grown in DMEM with 10% bovine serum, 
but the HUVEC were grown on gelatinized plates and in the presence of additional growth 
factors (bFGF and EGF). The untreated controls show similar levels of ^H-Thymidine 
5 incorporation for all three cell lines thus eliminating the concern that the differences in 

proliferation may be inherent. At the higher concentrations of vinblastine used (e.g. 100-400 
ng/ml) all three cell populations were strongly inhibited, especially HUVEC, In striking 
contrast, at the lowest concentrations (e.g. 0.78 ng/ml) vinblastine retained almost the same 
degree of inhibitory activity against HUVEC, whereas anti-proliferative activity against two 
10 tumor cell lines was not. The source of this differential sensitivity is not clear, but it should 
be noted that at least one of the tumor cell lines, SK-N-MC, is positive for multidrug 
resistance-associated protein (MRP). These in vitro findings suggest that the lowering of the 
usual maximum tolerated dose (MTD) used in the clinic may allow retention of good 
vinblastine activity against dividing endothelial cells present in tumors. 

15 

In vivo tumor growth assessment: Building on this in vitro difference in sensitivity to 
vinblastine, we went on to evaluate lower doses of * inblastine in an in vivo modeL using an 
increased dose frequency to maximize the endothe .:! injury. Xenografts of either SK-N-MC 
neuroepithelioma or .^iN.~:\-AS neuroblastoma cell lines were implanted subcutaneously in 

20 the flanks of 4-6 week old. CB-I7 SCID mice and grown to - 0.75 cm^ before initiation of 
treatment. The first treatment group, treated with DC 1 01 , an anti- Flkl receptor antibody 
shown previously to inhibit growth of different kinds of human xenografts in mice and in 
mouse tumor models (5), showed an anticipated effectiveness in inhibiting tumor growth, but, 
its effect was not sustained. The findings in the second treatment group (vinblastine alone), 

25 were even more surprising. This agent, traditionally thought to act by inhibiting tumor cell 
proliferation through inhibition of tubulin assembly, produced significant, albeit not 
sustained, regression of tumor growth even though used at subclinical low-dose,. This growth 
delay in the vinblastine group was further potentiated with the simultaneous treatment with 
the anti-flk-1 antibody, DClOl . The combination treatment induced an initial response 

30 comparable to the other treatment groups but then caused further, long term, tumor 
regression. To date, the mice in combination therapy group have not manifested any 
resistance to the treatment or recurrence of disease, despite almost seven months of 
continuous treatment. The mice remain healthy, with almost no evidence of tumor, except for 

24 



wo 01/54723 



PCT/USOl/02839 



a small, barely palpable remnant in one of the mice. 

Toxicity evaluation: Anti-vascular therapy would be expected to show minimal 
toxicity in the post-natal stage of development. To evaluate this aspect of DC 1 0 1 /vinblastine 
5 combination therapy, the health status of the mice was monitored. Weight was plotted at 

regular intervals and considered a surrogate for evaluation of systemic well being, anorexia, 
or failure to thrive. No significant differences in weights were seen between the four groups. 
The weight cun^'" of the DC 101 group parallels very closely that of the control group. The 
vinblastine group showed some weight gain retardation, but the differences never became 

10 significantly different from controls. Similarly, the toxicity profile in the combination 

treatment group was very similar to those in the single agent groups, with the exception of a 
transient episode of weight loss associated with diarrhea. The episode lasted approximately 
2-3 weeks and was unlikely to be due to the therapy as the mice recovered without 
interruption of treatment.Other usual signs of drug toxicity in mice such as ruffled fur, 

15 anorexia, cachexia, skin tenting (due to dehydration), skin ulcerations or toxic deaths, were 
not seen at the doses used in our experiments. Diarrhea, a common sign of vinblastine 
toxicity when doses of 10 mg/m- are used, was generally not observed, except for the above 
mentioned episode. 

20 Histopathologic analysis: To further elucidate the mechanisms involved in the tumor 

regression following treatment with vinblastine, DClOl, or the combined therapy, tissue 
histopathology assessment was undertaken. Cancer cells with high nuclear to cytoplasmic 
ratio form cuffs around central vessels, and apoptotic cells characterized by pyknotic nuclei 
and cytoplasmic blebbing, are only evident as a thin rim at the periphery of the cuffs. The 

25 nuclei of these cells stain strongly for terminal deoxynucleotidyl transferase (TUNEL) 

reactivity, as expected for cells undergoing apoptosis. Vinblastine alone or DC 101 treatment 
alone both show an increase in the width of the apoptotic rims, suggesting the cells most 
distal to the tumor vasculature are primarily affected, but a large percentage of viable tumor 
cells still survive in the center of the cuff. In contrast, histology of the combined therapy 

30 group, as would be predicted by the regression in tumor size in this treatment group at the 
time of analysis, shows overwhelming loss of both cell viability and pre-existing tumor 
architecture. There is a close similarity of the appearance of H/E and TUNEL stain. 
Interestingly, we observed signs of endothelial cell toxicity in all of the treatment groups. 

25 



wo 01/54723 



PCT/USOl/02839 



Rather than a typical single layer of flattened endothelial cells surrounding the vascular 
lumen in untreated group, we observed edema, and detachment from surrounding basement 
membrane and leading to complete vascular wall disintegration and tumor cell death. 

5 Tumor perfusion by assessment of intravascular fluorescence: To further explore the 

possibility that tumor regression induced with treatment using DC 101 and vinblastine was 
indeed due to the vascular injury, rather than a direct anti-tumor cell effect, we assessed 
txzmor perfusion directly by using a FITC-Dextran fluorescence method. Mice carrying 
established subcutaneous SK- N-AS human neuroblastoma xenografts (-0.75 cm^) were 

10 randomized into four groups and treated systemically with either saline control, DClOl , 

vinblastine or combination therapy for 10 days. FITC-Dextran was injected into the lateral 
tail vein and equilibrated throughout the vascular compartment. The majority of the blood- 
borne dextran, because of its 150 kDa size, remains intravascular, and despite some 
perivascular losses due to changes in vascular permeability and the possibility of interstitial 

15 hemorrhages, the fluorescence is reflective of the overall volume of blood passing through the 
tumor vasculature. Since our therapy is chronic in its nature, changes in intra- tumoral 
vascular/blood volume are likely to represent structural changes rather than transient fluxes in 
vascular permeability. By : e criteria DC 101 alone caused a 47 "'o decrease in tumor 
perfusion, whereas vinblasu. o aione resulted in a 41% decrease, and the combination of the 
20 two drugs resulted in 65% perfusion inhibition. Of interest is the appreciable difference in 
gross vascularity in the corresponding tumor specimens. 

Effects of chemotherapy treatments on in vivo angiogenesis: The direct assessment of 
tumor vasculature does not provide any clues as to whether the apparent vascular inhibition 

25 within the tumor is a primary cause or a secondary consequence of the tumor regression. 

Evidence for the former would provide support for the hypothesis that low-dose vinblastine 
treatment alone is potentially anti-angiogenic, and the extent of this anti-angiogenic effect 
may be ftirther enhanced by concurrent treatment with DC 101 . Again, the ratio between 
intra- and extra-vascular volume within the tumor could be also somewhat affected by 

30 transient changes in vascular permeability. To address these questions, we repeated the same 
fluorescence measurement using an in vivo Matrigel plug angiogenesis assay. Four treatment 
groups were treated with an identical therapeutic regimen as in the tumor perfusion 
experiment. The regression of vascularity in subcutaneously implanted Matrigel pellets was 

26 



wo 01/54723 



PCTAJSOl/02839 



quantitatively assessed by mea suring the fluorescence of circulating FITC-labeled dextran. 
DClOl administration inhibited bFGF induced vascularization to 50% of the positive control 
group, and vinblastine administration inhibited vascularization to 62,5% of the positive 
control group. There was again an enhanced effect with combination therapy, which reduced 
the Matrigel pellet fluorescence, and by inference vascularization to 29.2% of control, a level 
only marginally different to the negative control (Matrigel not supplemented with growth 
factors). 

Thus, large (0.75 cm^) established human neuroblastoma xenografts could be induced 
to completely regress with this combination strategy, whereas either agent alone caused only 
partial and temporary regressions with relapses observed in all animals treated at between SO- 
SO days after initiation of the individual therapy treatments. In striking contrast, a fully 
regressed state could be induced and maintained for as long as the combination therapy was 
maintained, which in our case was 200 days, in the absence of any significant toxicity, as 
assessed by lack of weight loss. No myelosuppression has been observed. 

The dose of vinblastine used in our experiments was in the range of 1 .5 mg/m^, every 
3 days, which is approximately 3 times the MTD of this drug in humans, and 1/16 - 1/20 of 
the MTD in mice, given the fact that the MTD of vinblastine in mice is 4-5 times higher than 
in humans. Using the Matrigel plug a>sa> we demonstrated that continuous low dose 
vinblastine administration can cause a direct anti-angiogenic effect in vivo. The combined 
effect with DC 101 was significant. 



27 



wo 01/54723 



PCT/USOl/02839 



What is claimed is: 

1. A method of treating or controlling an angiogenic dependent condition in a mammal 
comprising: 

administering an anti-angiogenic molecule and 

a chemotherapeutic agent to the mammal, in amounts and frequencies effective, in 
combination, to produce a regression or arrest of said condition while minimizing or 
preventing significant toxicity of the chemotherapeutic agent. 

2. The method of claim 1 , wherein the anti-angiogenic condition is selected from the 
group consisting of a neoplasm, a collagen-vascular disease or an auto-immune 
disease. 

3. The method of claim 2, wherein the neoplasm is a solid tumor. 

4. The method of claim 3, wherein the solid tumor is selected from the group consisting 
of breast carcinoma, lung carcinoma, prostate carcinoma, colon carcinoma, prostate 
carcinoma, ovarian carcinoma, neuroblastoma, central nervous system tumor, 
neuroblastoma, glioblastoma multiforme or melanoma. 

5. The method of claim K wherein the mammal is a human. 

6. The method of claim 1 , wherein the anti-angiogenic molecule inhibits or blocks the 
action of an vascular endothelium survival factor. 

7. The method of claim 6, wherein the vascular endothelial survival factor is selected 
from the group consisting of VEGF, VEGF receptor,avP3. avP3 receptor, Tie2/tek 
ligand, Tie2/tek, endoglin ligand, endoglin, neuropilin ligand, neuropilin, 
thrombospondin ligand, thrombospondin, PDGFa, PDGFa receptor, PDGFP, PDGFp 
receptor, aFGF, aFGF receptor, bFGF, bFGF receptor, TGpp, TGPp receptor, EGF, 
EGF receptor, angiostatin, angiostatin receptor, angiopoetin, angiopoeitin receptor, 
PLGF, PLGF receptor, VPF, or VPF receptor. 

8. The method of claim 6, wherein the vascular endothelial survival factor is a receptor. 

28 



wo 01/54723 



PCT/USOl/02839 



9. The method of claim 8, wherein the vascular endothelial survival factor is an 
angiogenesis growth factor receptor. 

10. The method of claim 9, wherein the angiogenesis growth factor receptor is a VEGF 
receptor. 

1 1 . The method of claim 10, wherein the VEGF receptor is selected from the group 
consisting of flk-l/KDR receptor, or flt-4 receptor. 

12. The method of claim 6, wherein the vascular endothelial survival factor is a ligand to 
a receptor. 

13. The method of claim 12, wherein the ligand is selected from the group consisting of 
VEGF, VEGF-B, VEGF-C, or VEGF-D. 

14. The method of claim 6, wherein the anti-angiogenic molecule is selected from the 
group consisting of an antibody, antibody fragment, small molecule or peptide. 

15. The method of claim 14, wherein the molecule is an antibody or fragment selected 
from the group consisting of mouse, rat, rabbit, chimeric, humanized or human 
antibody or fragment. 

16. The method of claim 6, wherein the anti-angiogenic molecule is IMC-lCl 1. 

17. The method of claim 16, wherein the IMC-lCl 1 is administered in a dose of from 
about 5 mg/m- to about 700 mg/m- from about daily to about every 7 days. 

1 8. The method of claim 1 7, wherein the IMC-1 C 1 1 is administered in a dose of from 
about 7.5 mg/m- to about 225 mg/m-, about twice per week. 

19. The method of claim 16, wherein the IMC-lCl 1 is administered at a dose and 
frequency sufficient to substantially saturate the VEGF receptor. 

29 



wo 01/54723 



PCT/USOl/02839 



20. The method of claim 6, wherein the anti-angiogenic molecule is administered in a 
dose and frequency sufficient to substantially saturate the target of the anti-angiogenic 
molecule. 

21 . The method of claim 1 , wherein the chemotherapeutic agent is selected from the 
group consisting of vinca alkaloid, camptothecan, taxane, or platinum analogue. 

22. The method of claim 21 , wherein the chemotherapeutic agent is selected from the 
group consisting of vincristine, vinblastine, vinorelbine, vindesine, paclitaxel, 
docetaxel, 5 FU, cisplatin, carboplatin, iranotecan, topotecan or cyclophosphamide. 

23. The method of claim 22, wherein the chemotherapeutic agent is administered at less 
than about 20% of the maximum tolerated dose. 

24. The method of claim 23, wherein the chemotherapeutic agent is administered at less 
than about 15% of the maximum tolerated dose. 

25. The method of claim 24, wherein the chemotherapeutic agent is administered at less 
than about 10% of the maximum tolerated dose. 

26. The method of claim 25, wherein the chemotherapeutic agent is administered at less 
than about 5% of the maximum tolerated dose. 

27. The method of claim 26, wherein the chemotherapeutic agent is administered at less 
than about 2% of the maximum tolerated dose. 

28. The method of claim 28, wherein the chemotherapeutic agent is administered at a dose 
intensity less than about 20% of the dose intensity of the chemotherapeutic agent 
when used in a conventional chemotherapeutic regimen, 

29. The method of claim 1 , wherein the chemotherapeutic agent is administered at a dose 
intensity less than about 1 0% of the dose intensity of the chemotherapeutic agent 

30 



wo 01/54723 



PCT/USOl/02839 



when used in a conventi 3nal chemotherapeutic regimen. 

30. The method of claim 29, v/herein the chemotherapeutic agent is administered at a dose 
intensity less than about 5% of the dose intensity of the chemotherapeutic agent when 
used in a conventional chemotherapeutic regimen. 

3 1 . The method of claim 22, wherein the chemotherapeutic agent is vinblastine 
administered in a dose from about 0.5 mg/m^ to about 3 mg/m^ from about once every 
3 days to about once every 7 days. 

32. The method of claim 1, wherein the chemotherapeutic agent is administered in a 
dosage and frequency that is of substantially equivalent efficacy to vinblastine in a 
dose from about 0.5 mg/m- to about 3 mg/m~ from about once every 3 days to about 
once every 7 days. 

33. The method of claim 1, wherein the chemotherapeutic agent is administered more 
frequently than about once every three weeks. 

34. The method of claim 33, wherein the chemotherapeutic agent is administered more 
frequently than about every seven days. 

35. A kit for treating an angiogenic dependent condition in a mammal comprising: 
an anti-angiogenic molecule; and, 

a chemotherapeutic agent, to be administered in amounts and frequencies effective, in 
combination, to produce a regression or arrest of the condition while minimizing or 
preventing significant toxicity of the chemotherapeutic agent, when administered in 
combination. 

36. The kit of claim 35, wherein the angiogenic dependent condition is selected from the 
group consisting of neoplasm, collagen-vascular disease or autoimmime disease, 

37. The kit of claim 36, wherein the neoplasm is a solid tumor. 



31 



wo 01/54723 



PCT/USOl/02839 



38. The kit of claim 37, wherein the solid tumor is selected from the group consisting of 
breast carcinoma, Ivmg carcinoma, prostate carcinoma, colon carcinoma, prostate 
carcinoma, ovarian carcinoma, neuroblastoma, central nervous system tumor, 
neuroblastoma, glioblastoma multiforme or melanoma. 

39. The kit of claim 35, wherein the mammal is a human. 

40. The kit of claim 35, wherein the anti-angiogenic molecule inhibits or blocks the action 
of a vascular endothelium survival factor. 

41. The kit of claim 40, wherein the vascular endothelial survival factor is selected from 
the group consisting of VEGF, VEGF receptor,avp3. cXyPs receptor, Tie2/tek ligand, 
Tie2/tek, endoglin ligand, endoglin, neuropilin ligand, neuropilin, thrombospondin 
ligand, thrombospondin, PDGFa, PDGFa receptor, PDGFp, PDGFp receptor, aFGF, 
aFGF receptor, bFGF, bFGF receptor, TGFp, TGPP receptor, EGF, EGF receptor, 
angiostatin, angiostatin receptor, angiopoetin, angiopoeitin receptor, PLGF, PLGF 
receptor, VPF, or VPF receptor. 

42. 1 le kit of claim 35, wherein the vascular endothelial surv ival factor is a receptor. 

43. The kit of claim 42, wherein the vascular endothelial survival factor is an 
angiogenesis growth factor receptor. 

44. The kit of claim 43, wherein the angiogenesis growth factor receptor is a VEGF 
receptor. 

45- The kit of claim 44, wherein the VEGF receptor is selected from the group consisting 
of flk- 1 /KDR receptor, or flt-4 receptor. 

46. The kit of claim 40, wherein the vascular endothelial survival factor is a ligand for a 
receptor. 



47. The kit of claim 46, wherein the ligand is selected from the group consisting of 

32 



wo 01/54723 



PCTAJSOl/02839 



VEGF, VEGF-B, VEGF-C, or VEGF-D. 

48. The kit of claim 40, wherein the anti-angiogenic molecule is selected from the group 
consisting of an antibody, antibody fragment, small molecule or peptide. 

49. The kit of claim 48, wherein the molecule is an antibody or fragment selected from 
the group consisting of mouse, rat, rabbit, chimeric, humanized or human antibody 
or fragn^'^nt. 

50. The kit of claim 48, wherein the antibody is IMC- 1 CI 1 . 

51. The kit of claim 50, wherein the IMC- 1 CI 1 is provided for administration in a dose of 
from about 5 mg/nr to about 700 mg/m~ about every 1 day to about every 7 days. 

52. The kit of claim 51, wherein the IMC-ICl 1 is provided for administration in a dose of 
from about 7.5 mg/m- to about 225 mg/m-, about twice per week. 

53. The kit of claim 50, wherein the IMC-lCl 1 is provided for administration at a dose 
and frequency sufficient to substantially saturate the VEGF receptor. 

54. The kit of claim 40, wherein the anti-angiogenic molecule is provided for 
administration in a dose and frequency sufficient to substantially saturate the target of 
the anti-angiogenic molecule. 

55. The kit of claim 35, wherein the chemotherapeutic agent is selected from the group 
consisting of a vinca alkaloid, a camptothecan, a taxane, or a platinum analogue. 

56. The kit of claim 55, wherein the chemotherapeutic agent is selected from the group 
consisting of vincristine, vinblastine, vinorelbine, vindesine, paclitaxel, docetaxel, 5 
FU, cisplatin, carboplatin, iranotecan, topotecan or cyclophosphamide. 

57. The kit of claim 35, wherein the chemotherapeutic agent is provided for 
administration at less than about 20% of the maximum tolerated dose. 

33 



wo 01/54723 



PCT/USOl/02839 



58. The kit of claim 57, wherein the chemotherapeutic agent is provided for 
administration at less than about 1 5% of the maximum tolerated dose. 

59. The kit of claim 58, wherein the chemotherapeutic agent is provided for 
administration at less than about 1 0% of the maximum tolerated dose. 

60. The kit of claim 59, wherein the chemotherapeutic agent is provided for 
administration at less than about 5% of the maximum tolerated dose. 

61. The kit of claim 60, wherein the chemotherapeutic agent is provided for 
administration at less than about 2% of the maximum tolerated dose. 

62. The kit of claim 35, wherein the chemotherapeutic agent is provided to be 
administered at a dose intensity less than about 20% of the dose intensity of the 
chemotherapeutic agent when used in a conventional chemotherapeutic regimen. 

63. The kit of claim 62. vherein the chemotherapeutic agent is provided to be 
administered at a dose intensity less than about 10% of the dose intensity of the 
chemotherapeutic agent when used in a conventional chemotherapeutic regimen. 

64. The kit of claim 63, wherein the chemotherapeutic agent is provided to be 
administered at a dose intensity less than about 5% of the dose intensity of the 
chemotherapeutic agent when used in a conventional chemotherapeutic regimen. 

65. The kit of claim 56, wherein the chemotherapeutic agent is vinblastine, provided for 
administration in a dose from about 0.5 mg/m~ to about 3 mg/nr from about once 
every 3 days to about once every 7 days. 

66. The kit of claim 35, wherein the chemotherapeutic agent is provided for 
administration at a dosage and frequency that is of substantially equivalent efficacy to 
vinblastine is a dose from about 0.5 mg/nr to about 3 mg/m^from about once every 3 
days to about once every 7 days. 

34 



wo 01/54723 



PCT/USOl/02839 



67. The kit of claim 35, wh*irein the chemotherapeutic agent is provided for 
administration more frequently than about every three weeks, 

5 68. The kit of claim 67, w^herein the chemotherapeutic agent is provided for 

administration more frequently than about every seven days. 



35 



wo 01/54723 



1/1 



PCTAJSOl/02839 



FIGURE 1 



Hindi I I 

GAACTT ATGGGATGGTCATGTATCATCCTTTTTCTAGTAGCAACTGCAACTGGAGTACAT 
MGWSCI IliFLVATATGVH 
leader 

TCACAGGTCAAGCTGCAGCAGTCTGGGGCAGAGCTTGTGGGGTCAGGGGCCTCAGTCAAA 
SQVKLQQSGAELVGSGASVK 
- > VH 

TTGTCCTGCACAACTTCTGGCTTCAACATTAAAGACTTCTATATGCACTGGGTGAAGCAG 
L S C T T S GFNIKDFYMH W V K Q 

CDR-Hl 

AGGCCTGAACAGGGCCTGGAGTGGATTGGATGGATTGATCCTGAGAATGGTGATTCTGAT 
RPEQGLEWIG WIDPENGDSD 

CDR-H2 

TATGCCCCGAAGTTCCAGGGCAAGGCCACCATGACTGCAGACTCATCCTCCAACACAGCC 

Y A P K F O G KATMTADSSSNTA 

TACCTGCAGCTCAGCAGCCTGACATCTGAGGACACTGCCGTCTATTACTGTAATGCATAC 
YLQLSSLTSEDTAVYYCNAY 

TATGGTGACTACGAAGGCTACTGGGGCCAAGGGACCACGGTCACCGTCTCCTCAGGTGAG 

Y G D Y E G Y WGQGTTVTVSS 

CDR-H3 

BamHI 
TGGATCC 



Hindu I 

AAGCTTATGGGATGGTCATGTATCATCCTTTTTCTAGTAGCAACTGCAACTGGAGTACAT 
MGWSCI ILFLVATATGVH 
>- leader 

TCAGACATCGAGCTCACTCAGTCTCCAGCAATCATGTCTGCATCTCCAGGGGAGAAGGTC 
SDIELTQSPAIMSASPGEKV 
► VL 

ACCATAACCTGCAGTGCCAGCTCAAGTGTAAGTTACATGCACTGGTTCCAGCAGAAGCCA 
T I T C SAS S SVS YMH W F Q Q K P 

CDR-Ll 

GGCACTTCTCCCAAACTCTGGATTTATAGCACATCCAACCTGGCTTCTGGAGTCCCTGCT 
GTSPKLWIY S T S N L A S G V P A 

CDR-L2 

CGCTTCAGTGGCAGTGGATCTGGGACCTCTTACTCTCTCACAATCAGCCGAATGGAGGCT 
RFSGSGSGTSYSLTISRMEA 

GAAGATGCTGCCACTTATTACTGCCAGCAAAGGAGTAGTTACCCATTCACGTTCGGCTCG 
EDAATYYC OORSSYPFT F G S 

CDR-L3 

BamHI 

GGGAC CAAGCTGGAAATAAAACGTGAGT GGATCC 
G T K L E I K 



wo 01/54723 



1 



PCT/USOl/02839 



SEQUENCE LISTING 

<110> Kerbel, Robert 

<120> Therapeutic Method for Reducing Angiogenesis 

<130> Sequence Listings 1-16 for 11245-19 

<140> Not yet as^^gned 
<141> 2000-03-20 

<150> US 60/178791 

<160> 16 

<170> Patentin Ver. 2.1 

<210> 1 
<211> 10 
<212> PRT 
<213> Mouse 

<400> 1 

Giy Phe Asn lie Lys Asp Phe Tyr Met His 
1 5 10 



<210> 2 
<211> 17 
<212> PRT 
<213> Mouse 

<400> 2 

Trp lie Asp Pro Glu Asn Giy 
1 5 



Giy 



AsD T/r Ala Pro Lys Phe Gin 
10 15 



<210> 3 
<211> 8 
<212> PRT 
<213> Mouse 

<400> 3 

Tyr Tyr Giy Asp Tyr Glu Giy Tyr 
1 5 



<210> 4 
<211> 10 
<212> PRT 
<213> Mouse 

<400> 4 

Ser Ala Ser Ser Ser Val Ser Tyr Met His 
1 5 10 



<210> 5 
<211> 7 
<212> PRT 
<213> Mouse 



wo 01/54723 



2 



PCT/USOl/02839 



<400> 5 

Ser Thr Ser Asn Leu Ala Ser 
1 5 



<210> 6 
<211> 9 
<212> PRT 
<213> Mouse 

<400> 6 

Gin Gin Arg Ser Ser Tyr Pro Phe Thr 
1 5 



<210> 7 
<211> 117 
<212> PRT 
<213> Mouse 

<400> 7 

Gin Val Lys Leu Gin Gin Ser Gly Ala Glu Leu Val Gly Ser Gly Ala 



15 10 1 



Ser Val Lys Leu Ser Cys Thr Thr Ser Gly Phe Asn lie Lys Asp Phe 

20 25 30 

Tyr Met His Trp Val Lys Gin Arg Pro Glu Gin Gly Leu Glu Trp lie 

35 40 45 

Gly Trp lie Asp Pro Glu Asn Gly Asp Ser Asp Tyr Ala Pro Lys Phe 
50 55 60 

"In Gly Lys Ala Thr Met Thr Ala Asp Ser Ser Ser Asn Thr Ala Tyr 
-5 70 ^ ^5 80 

.^In Leu Ser Ser Leu Thr Ser Glu Asp Tr;r Ala .''-i'^ Tyr Tyr Cys 

8 5 90 95 

Asn Ala Tyr Tyr Gly Asp Tyr Glu Gl'/ Tyr Tro Glw Gin Gly Thr Thr 

100 105 ^ ' ' 110 

Val Thr Val Ser Ser 
115 



<210> 8 
<211> 108 
<212> PRT 
<213> Mouse 

<400> 8 

Asp lie Glu Leu Thr Gin Ser Pro Ala lie Met Ser Ala Ser Pro Gly 
15 10 15 

Glu Lys Val Thr lie Thr Cys Ser Ala Ser Ser Ser Val Ser Tyr Met 

20 25 30 

His Trp Phe Gin Gin Lys Pro Gly Thr Ser Pro Lys Leu Trp lie Tyr 

35 40 45 

Ser Thr Ser Asn Leu Ala Ser Gly Val Pro Ala Arg Phe Ser Gly Ser 
50 55 60 

Gly Ser Gly Thr Ser Tyr Ser Leu Thr lie Ser Arg Met Glu Ala Glu 
65 70 75 80 



wo 01/54723 PCT/USOl/02839 



Asp Ala Ala Thr 

Phe Giy Ser Gly 

100 



Tyr Tyr Cys Gin 
85 

Thr Lys Leu Glu 



Gin Arg Ser Ser 
90 

lie Lys Arg Ala 
105 



Tyr Pro Phe Th - 

95 



<210> 9 
<211> 30 
<212> DNA 
<213> Mouse 

<400> 9 

ggcttcaaca ttaaagactt ctatatgcac 30 



<210> 10 
<211> 51 
<212> DNA 
<213> Mouse 

<400> 10 

tggattgatc ctgagaatgq tgattctgat tatgccccga agttccaggg c 51 



<210> 11 
<211> 24 
<212> DNA 
<213> Mouse 

<400> 11 

tactatggtg actacgaagg ctac 24 



<21C> 12 

<^12> DNA 
<213> Mouse 

< 4 0 0 > 12 

agtgccagct caagtgtaag ttacatgcac 30 



<210> 13 
<211> 21 
<212> DNA 
<213> Mouse 

<400> 13 

agcacatcca acctggcttc t 21 



<210> 14 
<211> 27 
<212> DNA 
<213> Mouse 

<400> 14 

cagcaaagga gtagttaccc attcacg 27 



<210> 15 
<211> 351 
<212> DNA 
<213> Mouse 



wo 01/54723 



4 



PCT/USOl/02839 



<400> 15 

caggtcaagc tgcagcagtc tggggcagag cttgtggggt caggggcctc agtcaaattg 60 

tcctgcacaa cttctggctt caacattaaa gacttctata tgcactgggt gaagcagagg 120 

cctgaacagg gcctggagtg gattggatgg attgatcctg agaatggtga ttctgattat 180 

gccccgaagt tccagggcaa ggccaccatg actgcagact catcctccaa cacagcctac 240 

ctgcagctca gcagcctgac atctgaggac actgccgtct attactgtaa tgcatactat 300 

ggtgactacg aaggctactg gggccaaggg accacggtca ccgcctcctc a 351 

<210> 16 
<211> 324 
<212> DNA 
<213> Mouse 

<400> 16 

gacatcgagc tcactcagtc tccagcaatc atgtctgcat ctccagggga gaaggtcacc 60 
ataacctgca gtgccagctc aagtgtaagt tacatgcact ggttccagca gaagccaggc 120 
acttctccca aactctggat ttatagcaca tccaacctgg cttctggagt ccctgctcgc 180 
ttcagtggca gtggatctgg gacctcttac tctctcacaa tcagccgaat ggaggctgaa 240 
gatgctgcca cttattactg ccagcaaagg agtagttacc cattcacgtt cggctcgggg 300 
accaagctgg aaataaaacg ggcg 324 



INTERNATIONAL SEARCH REPORT 



International application No. 



PCT/USOl/02839 



A. CXASSIFICATION OF SUBJECT MATTER 

IPC(7) A61K 39/395, 39/00, 39/38; GOIN 33/53 

US CL 424/ 130.1, 138.1, 143,1, 184.1; 435/975 

According to International Patent Classification (IPC) or to both national classification and IPC 



B. 



FIELDS SEARCHED 



M inimum documentation searched (classification system followed by classification symbols) 
U.S. : 424/ 130.1, 138.1, 143.1. 184.1; 435/975 



Documentation searched other than minimum docimientation to the extent that such documents are included in the fields searched 



Electronic data base consulted during the international search (name of data base and, where practicable, search terms used) 
STN, WEST, MEDLINE, 



C. DOCUMENTS CONSIDERED TO BE RELEVANT 



Category 



Citation of document, with indication, where appropriate, of the relevant passages 



Relevant to claim No. 



US 5.840,301 A (ROCKWELL et al.) 24 November 1998 (24. IL 1998), entire article. 



Y 
X 

Y 
Y 



US 5.874,542 A (ROCKWELL et al.) 23 February 1999 (23.02.1999), entire article. 



Database PROMT, Accession Number 1999:838616, ImCLONE SYSTEMS INC. 
ImClone Files Investigational New Drug Application for Anti-Angiogenesis. Business 
Wire. 15 December 1999 (15.12. 1999), entire article. 

DEVTTA et al. Cancer: Principles & Practice of Oncology, 5th edition, 1997, pages 
333, 405,418,432. 



1-15,20-22 

35-49, 54-56 
1-15,20-22 

35-49, 54-56 
16-19,50-53 



23-34,57-68 



□ 



Further documents are listed in the continuation of Box C. 



□ 



See patent family annex. 



* special categories of cited dDcumenu: 

**A" document derming tlie general state of the an which is not considered to be 
<^ particular relevance 

"E" earlier application or patem published on or after the international filing date 

*L** document which may throw doubts on priority claim(s) or which is cited to 
est^>Ush the publication date aC anodier citation or other special reason (as 
specified) 

"O** documeni referrmg to an oral disclosure, use, exhilHtion or other means 

'P" document puUished prior to the international Tiling date but later than the 
priority date claimed 



"X' 



later document pobiisbed after the international fdiog date or priority 
date and not in conflict with the application but cited to understand the 
principle or theory underlying the invention 

document of partictilar relevance; the claimed invention cannot be 
considered nofve! or cannot be considered to involve an inventive step 
when the document is taken alone 

docmnent of particnlar relevance; the claimed invention cannol be 
considered to involve an inventive step when the document is 
combined with one or more other such documents, such cooabtnaiion 
being obvioas to a person sldlled m the art 

documeni member of the same patent family 



Date of the actual completion of the international search 
08 March 2001 (08.03.2001) 



Name and mailing address of the ISA/US 

Commissioner of Patents and Trademarks 
Boot PCX 

Washmgiott, D.C. 20231 

Facsimile No. (703)305-3230 



Date of mailing of the international search report 

28 JUN20O1 





Telephone No. 703-308-0196 



Form PCT/ISA/210 (second sheet) (July 1998)